101
|
Garcia-Cuellar MP, Steger J, Füller E, Hetzner K, Slany RK. Pbx3 and Meis1 cooperate through multiple mechanisms to support Hox-induced murine leukemia. Haematologica 2015; 100:905-13. [PMID: 25911551 DOI: 10.3324/haematol.2015.124032] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 04/17/2015] [Indexed: 12/18/2022] Open
Abstract
Hox homeobox transcription factors drive leukemogenesis efficiently only in the presence of Meis or Pbx proteins. Here we show that Pbx3 and Meis1 need to dimerize to support Hox-induced leukemia and we analyze the molecular details of this cooperation. In the absence of Pbx3, Meis1 was highly unstable. As shown by a deletion analysis Meis1 degradation was contingent on a motif coinciding with the Pbx-binding domain. Either deletion of this sequence or binding to Pbx3 prolonged the half-life of Meis1 by preventing its ubiquitination. Meis1 break-down could also be blocked by inhibition of the ubiquitin proteasome system, indicating tight post-transcriptional control. In addition, Meis1 and Pbx3 cooperated genetically as overexpression of Pbx3 induced endogenous Meis1 transcription. These functional interactions translated into in vivo activity. Blocking Meis1/Pbx3 dimerization abrogated the ability to enhance proliferation and colony-forming cell numbers in primary cells transformed by Hoxa9. Furthermore, expression of Meis1 target genes Flt3 and Trib2 was dependent on Pbx3/Meis1 dimerization. This correlated with the requirement of Meis1 to bind Pbx3 in order to form high affinity DNA/Hoxa9/Meis1/Pbx3 complexes in vitro. Finally, kinetics and severity of disease in transplantation assays indicated that Pbx3/Meis1 dimers are rate-limiting factors for Hoxa9-induced leukemia.
Collapse
Affiliation(s)
| | - Julia Steger
- Department of Genetics, Friedrich-Alexander-University, Erlangen, Germany
| | - Elisa Füller
- Department of Genetics, Friedrich-Alexander-University, Erlangen, Germany
| | - Katrin Hetzner
- Department of Genetics, Friedrich-Alexander-University, Erlangen, Germany
| | - Robert K Slany
- Department of Genetics, Friedrich-Alexander-University, Erlangen, Germany
| |
Collapse
|
102
|
Friedman AD. C/EBPα in normal and malignant myelopoiesis. Int J Hematol 2015; 101:330-41. [PMID: 25753223 DOI: 10.1007/s12185-015-1764-6] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Revised: 02/18/2015] [Accepted: 02/19/2015] [Indexed: 12/22/2022]
Abstract
CCAAT/enhancer binding protein α (C/EBPα) dimerizes via its leucine zipper (LZ) domain to bind DNA via its basic region and activate transcription via N-terminal trans-activation domains. The activity of C/EBPα is modulated by several serine/threonine kinases and via sumoylation, its gene is activated by RUNX1 and additional transcription factors, its mRNA stability is modified by miRNAs, and its mRNA is subject to translation control that affects AUG selection. In addition to inducing differentiation, C/EBPα inhibits cell cycle progression and apoptosis. Within hematopoiesis, C/EBPα levels increase as long-term stem cells progress to granulocyte-monocyte progenitors (GMP). Absence of C/EBPα prevents GMP formation, and higher levels are required for granulopoiesis compared to monopoiesis. C/EBPα interacts with AP-1 proteins to bind hybrid DNA elements during monopoiesis, and induction of Gfi-1, C/EBPε, KLF5, and miR-223 by C/EBPα enables granulopoiesis. The CEBPA ORF is mutated in approximately 10 % of acute myeloid leukemias (AML), leading to expression of N-terminally truncated C/EBPαp30 and C-terminal, in-frame C/EBPαLZ variants, which inhibit C/EBPα activities but also play additional roles during myeloid transformation. RUNX1 mutation, CEBPA promoter methylation, Trib1 or Trib2-mediated C/EBPαp42 degradation, and signaling pathways leading to C/EBPα serine 21 phosphorylation reduce C/EBPα expression or activity in additional AML cases.
Collapse
Affiliation(s)
- Alan D Friedman
- Division of Pediatric Oncology, Johns Hopkins University, Cancer Research Building I, Room 253, 1650 Orleans Street, Baltimore, MD, 21231, USA,
| |
Collapse
|
103
|
MEIS1 regulates an HLF-oxidative stress axis in MLL-fusion gene leukemia. Blood 2015; 125:2544-52. [PMID: 25740828 DOI: 10.1182/blood-2014-09-599258] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 02/18/2015] [Indexed: 01/15/2023] Open
Abstract
Leukemias with MLL translocations are often found in infants and are associated with poor outcomes. The pathogenesis of MLL-fusion leukemias has been linked to upregulation of HOX/MEIS1 genes. The functions of the Hox/Meis1 complex in leukemia, however, remain elusive. Here, we used inducible Meis1-knockout mice coupled with MLL-AF9 knockin mice to decipher the mechanistic role of Meis1 in established MLL leukemia. We demonstrate that Meis1 is essential for maintenance of established leukemia. In addition, in both the murine model and human leukemia cells, we found that Meis1 loss led to increased oxidative stress, oxygen flux, and apoptosis. Gene expression and chromatin immunoprecipitation studies revealed hepatic leukemia factor (HLF) as a target gene of Meis1. Hypoxia or HLF expression reversed the oxidative stress, rescuing leukemia development in Meis1-deficient cells. Thus, the leukemia-promoting properties of Meis1 are at least partly mediated by a low-oxidative state, aided by HLF. These results suggest that stimulants of oxidative metabolism could have therapeutic potential in leukemia treatment.
Collapse
|
104
|
Ebina W, Rossi DJ. Transcription factor-mediated reprogramming toward hematopoietic stem cells. EMBO J 2015; 34:694-709. [PMID: 25712209 DOI: 10.15252/embj.201490804] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
De novo generation of human hematopoietic stem cells (HSCs) from renewable cell types has been a long sought-after but elusive goal in regenerative medicine. Paralleling efforts to guide pluripotent stem cell differentiation by manipulating developmental cues, substantial progress has been made recently toward HSC generation via combinatorial transcription factor (TF)-mediated fate conversion, a paradigm established by Yamanaka's induction of pluripotency in somatic cells by mere four TFs. This review will integrate the recently reported strategies to directly convert a variety of starting cell types toward HSCs in the context of hematopoietic transcriptional regulation and discuss how these findings could be further developed toward the ultimate generation of therapeutic human HSCs.
Collapse
Affiliation(s)
- Wataru Ebina
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA Program in Cellular and Molecular Medicine, Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| | - Derrick J Rossi
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA Program in Cellular and Molecular Medicine, Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA Department of Pediatrics, Harvard Medical School, Boston, MA, USA Harvard Stem Cell Institute, Cambridge, MA, USA
| |
Collapse
|
105
|
Li BE, Ernst P. Two decades of leukemia oncoprotein epistasis: the MLL1 paradigm for epigenetic deregulation in leukemia. Exp Hematol 2014; 42:995-1012. [PMID: 25264566 PMCID: PMC4307938 DOI: 10.1016/j.exphem.2014.09.006] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 09/16/2014] [Indexed: 12/12/2022]
Abstract
MLL1, located on human chromosome 11, is disrupted in distinct recurrent chromosomal translocations in several leukemia subsets. Studying the MLL1 gene and its oncogenic variants has provided a paradigm for understanding cancer initiation and maintenance through aberrant epigenetic gene regulation. Here we review the historical development of model systems to recapitulate oncogenic MLL1-rearrangement (MLL-r) alleles encoding mixed-lineage leukemia fusion proteins (MLL-FPs) or internal gene rearrangement products. These largely mouse and human cell/xenograft systems have been generated and used to understand how MLL-r alleles affect diverse pathways to result in a highly penetrant, drug-resistant leukemia. The particular features of the animal models influenced the conclusions of mechanisms of transformation. We discuss significant downstream enablers, inhibitors, effectors, and collaborators of MLL-r leukemia, including molecules that directly interact with MLL-FPs and endogenous mixed-lineage leukemia protein, direct target genes of MLL-FPs, and other pathways that have proven to be influential in supporting or suppressing the leukemogenic activity of MLL-FPs. The use of animal models has been complemented with patient sample, genome-wide analyses to delineate the important genomic and epigenomic changes that occur in distinct subsets of MLL-r leukemia. Collectively, these studies have resulted in rapid progress toward developing new strategies for targeting MLL-r leukemia and general cell-biological principles that may broadly inform targeting aberrant epigenetic regulators in other cancers.
Collapse
Affiliation(s)
- Bin E Li
- Department of Genetics, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Patricia Ernst
- Department of Genetics, Geisel School of Medicine at Dartmouth, Hanover, NH, USA; Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA; Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Hanover, NH, USA; Department of Pediatrics Hematology/Oncology/BMT, University of Colorado Anschutz Medical Center, Aurora, CO, USA.
| |
Collapse
|
106
|
Chopra M, Bohlander SK. Disturbing the histone code in leukemia: translocations and mutations affecting histone methyl transferases. Cancer Genet 2014; 208:192-205. [PMID: 25592767 DOI: 10.1016/j.cancergen.2014.10.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 10/01/2014] [Accepted: 10/14/2014] [Indexed: 10/24/2022]
Abstract
Leukemia is characterized by increased numbers of blasts originating from transformed early hematopoietic stem and progenitor cells. Genetic alterations are widely recognized as the main drivers of oncogenic transformation. Of considerable interest are mutations affecting the writers of epigenetic marks. In this review, we focus on histone methyltransferases--enzymes that catalyze the methylation of lysine residues in core histones. Histone methylation is a tightly controlled mechanism that is responsible for both activating as well as repressing gene expression in a site-specific manner, depending on which lysine residue is methylated. Histone methyltransferases, including MLL1, DOT1L, EZH2, and SETD2 are recurrently deregulated in human leukemia, either directly by gene mutations or balanced translocations, or indirectly as components of protein complexes that are disturbed in leukemia due to alterations of the other components in these complexes. Several small molecule inhibitors of histone methyltransferases are currently being clinically evaluated for their therapeutic potential in human leukemia. These drugs reverse some of the adverse effects of aberrant histone methylation, and can induce differentiation and cell death in leukemic blasts.
Collapse
Affiliation(s)
- Martin Chopra
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Stefan K Bohlander
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand.
| |
Collapse
|
107
|
Insulin-like growth factor 1 is a direct HOXA9 target important for hematopoietic transformation. Leukemia 2014; 29:901-8. [DOI: 10.1038/leu.2014.287] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 09/12/2014] [Accepted: 09/22/2014] [Indexed: 12/18/2022]
|
108
|
Brumatti G, Salmanidis M, Kok CH, Bilardi RA, Sandow JJ, Silke N, Mason K, Visser J, Jabbour AM, Glaser SP, Okamoto T, Bouillet P, D'Andrea RJ, Ekert PG. HoxA9 regulated Bcl-2 expression mediates survival of myeloid progenitors and the severity of HoxA9-dependent leukemia. Oncotarget 2014; 4:1933-47. [PMID: 24177192 PMCID: PMC3875760 DOI: 10.18632/oncotarget.1306] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Deregulated expression of Hox genes such as HoxA9 is associated with development of myeloproliferative disorders and leukemia and indicates a poor prognosis. To investigate the molecular mechanisms by which HoxA9 promotes immortalization of hematopoietic cells, we generated growth factor dependent myeloid cells in which HoxA9 expression is regulated by administration of 4-hydroxy-tamoxifen. Maintenance of HoxA9 overexpression is required for continued cell survival and proliferation, even in the presence of growth factors. We show for the first time that maintenance of Bcl-2 expression is critical for HoxA9-dependent immortalization and influences the latency of HoxA9-dependent leukemia. Hematopoietic cells lacking Bcl-2 were not immortalized by HoxA9 in vitro. Furthermore, deletion of Bcl-2 delayed the onset and reduced the severity of HoxA9/Meis1 and MLL-AF9 leukemias. This is the first description of a molecular link between HoxA9 and the regulation of Bcl-2 family members in acute myeloid leukemia.
Collapse
Affiliation(s)
- Gabriela Brumatti
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
109
|
Spivak JL, Considine M, Williams DM, Talbot CC, Rogers O, Moliterno AR, Jie C, Ochs MF. Two clinical phenotypes in polycythemia vera. N Engl J Med 2014; 371:808-17. [PMID: 25162887 PMCID: PMC4211877 DOI: 10.1056/nejmoa1403141] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Polycythemia vera is the ultimate phenotypic consequence of the V617F mutation in Janus kinase 2 (encoded by JAK2), but the extent to which this mutation influences the behavior of the involved CD34+ hematopoietic stem cells is unknown. METHODS We analyzed gene expression in CD34+ peripheral-blood cells from 19 patients with polycythemia vera, using oligonucleotide microarray technology after correcting for potential confounding by sex, since the phenotypic features of the disease differ between men and women. RESULTS Men with polycythemia vera had twice as many up-regulated or down-regulated genes as women with polycythemia vera, in a comparison of gene expression in the patients and in healthy persons of the same sex, but there were 102 genes with differential regulation that was concordant in men and women. When these genes were used for class discovery by means of unsupervised hierarchical clustering, the 19 patients could be divided into two groups that did not differ significantly with respect to age, neutrophil JAK2 V617F allele burden, white-cell count, platelet count, or clonal dominance. However, they did differ significantly with respect to disease duration; hemoglobin level; frequency of thromboembolic events, palpable splenomegaly, and splenectomy; chemotherapy exposure; leukemic transformation; and survival. The unsupervised clustering was confirmed by a supervised approach with the use of a top-scoring-pair classifier that segregated the 19 patients into the same two phenotypic groups with 100% accuracy. CONCLUSIONS Removing sex as a potential confounder, we identified an accurate molecular method for classifying patients with polycythemia vera according to disease behavior, independently of their JAK2 V617F allele burden, and identified previously unrecognized molecular pathways in polycythemia vera outside the canonical JAK2 pathway that may be amenable to targeted therapy. (Funded by the Department of Defense and the National Institutes of Health.).
Collapse
Affiliation(s)
- Jerry L Spivak
- From the Division of Hematology, Department of Medicine (J.L.S., D.M.W., O.R., A.R.M.), Division of Biostatistics and Bioinformatics, Sidney Kimmel Comprehensive Cancer Center (M.C.), and the Basic Science Institute (C.C.T.), Johns Hopkins University School of Medicine, Baltimore; the Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago (C.J.); and the Department of Mathematics and Statistics, College of New Jersey, Ewing (M.F.O.)
| | | | | | | | | | | | | | | |
Collapse
|
110
|
Kettyle LMJ, Liberante FG, Thompson A. Rational drug repurposing using sscMap analysis in a HOX-TALE model of leukemia. Methods Mol Biol 2014; 1196:349-70. [PMID: 25151174 DOI: 10.1007/978-1-4939-1242-1_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Abstract
Drug discovery and development are often hampered by lack of target identification and clinical tractability. Repurposing of approved drugs to life-threatening diseases such as leukemia is emerging as a promising alternative approach. Connectivity mapping systems link approved drugs with disease-related gene signatures. Relevant preclinical models provide essential tools for system validation and proof-of-concept studies. Herein we describe procedures aimed at generating disease-based gene signatures and applying them to established cross-referencing databases of potential candidate drugs. As a proof of principle, we present the identification of Entinostat as a candidate drug for the treatment of HOX-TALE-related leukemia.
Collapse
Affiliation(s)
- Laura M J Kettyle
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, 97 Lisburn Road, Belfast, Northern Ireland, BT9 7BL, UK
| | | | | |
Collapse
|
111
|
Yui MA, Rothenberg EV. Developmental gene networks: a triathlon on the course to T cell identity. Nat Rev Immunol 2014; 14:529-45. [PMID: 25060579 PMCID: PMC4153685 DOI: 10.1038/nri3702] [Citation(s) in RCA: 248] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cells acquire their ultimate identities by activating combinations of transcription factors that initiate and sustain expression of the appropriate cell type-specific genes. T cell development depends on the progression of progenitor cells through three major phases, each of which is associated with distinct transcription factor ensembles that control the recruitment of these cells to the thymus, their proliferation, lineage commitment and responsiveness to T cell receptor signals, all before the allocation of cells to particular effector programmes. All three phases are essential for proper T cell development, as are the mechanisms that determine the boundaries between each phase. Cells that fail to shut off one set of regulators before the next gene network phase is activated are predisposed to leukaemic transformation.
Collapse
Affiliation(s)
- Mary A Yui
- Division of Biology 156-29, California Institute of Technology, Pasadena, California 91125, USA
| | - Ellen V Rothenberg
- Division of Biology 156-29, California Institute of Technology, Pasadena, California 91125, USA
| |
Collapse
|
112
|
Ma H, Mallampati S, Lu Y, Sun B, Wang E, Leng X, Gong Y, Shen H, Yin CC, Jones D, Amin HM, You MJ, Zweidler-McKay P, Ma Y, Kantarjian HM, Arlinghaus RB, Glassman A, Sun X. The Sox4/Tcf7l1 axis promotes progression of BCR-ABL-positive acute lymphoblastic leukemia. Haematologica 2014; 99:1591-8. [PMID: 24997151 DOI: 10.3324/haematol.2014.104695] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The transcription factor Sox4 plays an indispensable role in the development of early progenitor B cells from hematopoietic stem cells. However, its role in B-cell acute lymphoblastic leukemia, a malignant counterpart of normal progenitor B cells, is not fully understood. Here we show that SOX4 is highly expressed in human acute lymphoblastic leukemia cells. To systematically study the function of Sox4 in acute lymphoblastic leukemia, we established a genetically defined mouse leukemia model by transforming progenitor B cells carrying a floxed Sox4 allele and inducing deletion of the allele by the self-excising Cre recombinase. This model allowed us to work with two groups of leukemic cells that had either one copy or both copies of Sox4 deleted. We found that depletion of Sox4 in transformed cells in vitro reduced cell growth in vitro and the progression of leukemia in vivo. Moreover, depletion of Sox4 in leukemic cells in vivo prolonged the survival of the mice, suggesting that it could be a potential target in acute lymphoblastic leukemia therapy. Our microarray and bioChIP studies revealed that Tcf7l1 was the key gene directly regulated by Sox4. Knockdown of Tcf7l1 reduced cell proliferation, just as did knockout of Sox4, and ectopic expression of Tcf7l1 could reverse the effect of Sox4 knockout on cell proliferation. These data suggest that Sox4 and Tcf7l1 form a functional axis that promotes the progression of BCR-ABL-positive acute lymphoblastic leukemia.
Collapse
Affiliation(s)
- Haiqing Ma
- Department of Laboratory Medicine and the Center for Stem Cell and Developmental Biology, The University of Texas MD Anderson Cancer Center (MDACC), Houston, TX, USA Department of Oncology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Saradhi Mallampati
- Department of Laboratory Medicine and the Center for Stem Cell and Developmental Biology, The University of Texas MD Anderson Cancer Center (MDACC), Houston, TX, USA
| | - Yue Lu
- Department of Molecular Carcinogenesis, The University of Texas MDACC, Houston, TX, USA
| | - Baohua Sun
- Department of Laboratory Medicine and the Center for Stem Cell and Developmental Biology, The University of Texas MD Anderson Cancer Center (MDACC), Houston, TX, USA
| | - Enze Wang
- Department of Laboratory Medicine and the Center for Stem Cell and Developmental Biology, The University of Texas MD Anderson Cancer Center (MDACC), Houston, TX, USA
| | - Xiaohong Leng
- Department of Translational Molecular Pathology, The University of Texas MDACC, Houston, TX, USA
| | - Yun Gong
- Department of Pathology, The University of Texas MDACC, Houston, TX, USA
| | - Haifa Shen
- Department of Nanomedicine, Houston Methodist Research Institute, TX, and Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY, USA
| | - C Cameron Yin
- Department of Hematopathology, The University of Texas MDACC, Houston, TX, USA
| | - Dan Jones
- School of Health Sciences, The University of Texas MDACC, Houston, TX, USA
| | - Hesham M Amin
- Department of Hematopathology, The University of Texas MDACC, Houston, TX, USA
| | - M James You
- Department of Hematopathology, The University of Texas MDACC, Houston, TX, USA
| | | | - Yupo Ma
- Department of Pathology, Stony Brook University Medical Center, Stony Brook, NY, USA
| | | | - Ralph B Arlinghaus
- Department of Translational Molecular Pathology, The University of Texas MDACC, Houston, TX, USA
| | - Armand Glassman
- Department of Microbiology and Immunology, The Medical University of South Carolina, Charleston, and Department of Pathology & Laboratory Medicine, The University of Texas Houston Health Science Center, Houston, TX, USA
| | - Xiaoping Sun
- Department of Laboratory Medicine and the Center for Stem Cell and Developmental Biology, The University of Texas MD Anderson Cancer Center (MDACC), Houston, TX, USA
| |
Collapse
|
113
|
C/EBPα is an essential collaborator in Hoxa9/Meis1-mediated leukemogenesis. Proc Natl Acad Sci U S A 2014; 111:9899-904. [PMID: 24958854 DOI: 10.1073/pnas.1402238111] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Homeobox A9 (HOXA9) is a homeodomain-containing transcription factor that plays a key role in hematopoietic stem cell expansion and is commonly deregulated in human acute leukemias. A variety of upstream genetic alterations in acute myeloid leukemia (AML) lead to overexpression of HOXA9, almost always in association with overexpression of its cofactor meis homeobox 1 (MEIS1) . A wide range of data suggests that HOXA9 and MEIS1 play a synergistic causative role in AML, although the molecular mechanisms leading to transformation by HOXA9 and MEIS1 remain elusive. In this study, we identify CCAAT/enhancer binding protein alpha (C/EBPα) as a critical collaborator required for Hoxa9/Meis1-mediated leukemogenesis. We show that C/EBPα is required for the proliferation of Hoxa9/Meis1-transformed cells in culture and that loss of C/EBPα greatly improves survival in both primary and secondary murine models of Hoxa9/Meis1-induced leukemia. Over 50% of Hoxa9 genome-wide binding sites are cobound by C/EBPα, which coregulates a number of downstream target genes involved in the regulation of cell proliferation and differentiation. Finally, we show that Hoxa9 represses the locus of the cyclin-dependent kinase inhibitors Cdkn2a/b in concert with C/EBPα to overcome a block in G1 cell cycle progression. Together, our results suggest a previously unidentified role for C/EBPα in maintaining the proliferation required for Hoxa9/Meis1-mediated leukemogenesis.
Collapse
|
114
|
Wang L, Li Y, Yang X, Yuan H, Li X, Qi M, Chang YWY, Wang C, Fu W, Yang M, Zhang J, Han B. ERG-SOX4 interaction promotes epithelial-mesenchymal transition in prostate cancer cells. Prostate 2014; 74:647-58. [PMID: 24435928 DOI: 10.1002/pros.22783] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Accepted: 12/31/2013] [Indexed: 12/27/2022]
Abstract
BACKGROUND Approximately 50% of prostate cancer (PCa) patients in Western countries harbor ERG rearrangement with concurrent ERG overexpression. Overexpression of SOX4 has been shown to play important roles in multiple cancers including PCa. However, the link between these two critical genetic aberrations was unclear. METHODS Fluorescence in situ hybridization and immunohistochemistry were utilized to detect ERG rearrangement and SOX4 expression. Cellular function was evaluated by transwell, wound healing assays, and cell adhesion assay, respectively. Interaction between ERG and SOX4 was arrayed by co-immunoprecipitation, Real-time PCR, Western blot, and siRNA. Direct binding of ERG to the promoter of SOX4, as well as epigenetic modifications of their promoters after TGF-β1 treatment was monitored by chromatin immunoprecipitation. RESULTS ERG regulated SOX4 expression via binding to its promoter. Silencing both of them showed duplicate effects on restoring the epithelial characteristics, increasing cellular adhesion and decreasing capacity of cellular migration and invasion. ERG and SOX4 have cooperative roles in TGF-β1-induced epithelial to mesenchymal transition (EMT) process. In addition, TGF-β1 stimulation increased levels of chromatin marks associated with active genes (H3K4me3, H416ac), and decreased levels of repressive marks (H3K27me3) at their promoters. 5-aza and TSA treatment changed expressions of ERG and SOX4. Clinically, overexpression of SOX4 is associated with ERG rearrangement status in PCa and ERG+/SOX4+ defined a subset of PCa patients with poor prognosis. CONCLUSION Our findings define a key role for ERG/SOX4 in the development of a subset of PCa and highlight the clinical importance of identifying molecularly defined tumor subgroups.
Collapse
Affiliation(s)
- Lin Wang
- Department of Pathology, Shandong University Medical School, Jinan, China; Research Center for Medicinal Biotechnology, Shandong Academy of Medicinal Sciences, Jinan, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
115
|
Palma CA, Al Sheikha D, Lim TK, Bryant A, Vu TT, Jayaswal V, Ma DDF. MicroRNA-155 as an inducer of apoptosis and cell differentiation in Acute Myeloid Leukaemia. Mol Cancer 2014; 13:79. [PMID: 24708856 PMCID: PMC4021368 DOI: 10.1186/1476-4598-13-79] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Accepted: 03/27/2014] [Indexed: 12/17/2022] Open
Abstract
Background Acute myeloid leukaemia (AML) is characterised by the halt in maturation of myeloid progenitor cells, combined with uncontrolled proliferation and abnormal survival, leading to the accumulation of immature blasts. In many subtypes of AML the underlying causative genetic insults are not fully described. MicroRNAs are known to be dysregulated during oncogenesis. Overexpression of miR-155 is associated with some cancers, including haematological malignancies, and it has been postulated that miR-155 has an oncogenic role. This study investigated the effects of modulating miR-155 expression in human AML cells, and its mechanism of action. Results Analysis of miR-155 expression patterns in AML patients found that Fms-like tyrosine kinase 3 (FLT3)-wildtype AML has the same expression level as normal bone marrow, with increased expression restricted to AML with the FLT3-ITD mutation. Induction of apoptosis by cytarabine arabinoside or myelomonocytic differentiation by 1,23-dihydroxyvitaminD3 in FLT3-wildtype AML cells led to upregulated miR-155 expression. Knockdown of miR-155 by locked nucleic acid antisense oligonucleotides in the FLT3-wildtype AML cells conferred resistance to cytarabine arabinoside induced apoptosis and suppressed the ability of cells to differentiate. Ectopic expression of miR-155 in FLT3-wildtype AML cells led to a significant gain of myelomonocytic markers (CD11b, CD14 and CD15), increase in apoptosis (AnnexinV binding), decrease in cell growth and clonogenic capacity. In silico target prediction identified a number of putative miR-155 target genes, and the expression changes of key transcription regulators of myeloid differentiation and apoptosis (MEIS1, GF1, cMYC, JARID2, cJUN, FOS, CTNNB1 and TRIB2) were confirmed by PCR. Assessment of expression of apoptosis-related proteins demonstrated a marked increase in cleaved caspase-3 expression confirming activation of the apoptosis cascade. Conclusions This study provides evidence for an anti-leukaemic role for miR-155 in human FLT3-wildtype AML, by inducing cell apoptosis and myelomonocytic differentiation, which is in contrast to its previously hypothesized role as an oncogene. This highlights the complexity of gene regulation by microRNAs that may have tumour repressor or oncogenic effects depending on disease context or tissue type.
Collapse
Affiliation(s)
| | | | | | | | | | | | - David D F Ma
- Blood, Stem Cells and Cancer Research, St Vincent's Centre for Applied Medical Research, St Vincent's Hospital, Sydney, Australia.
| |
Collapse
|
116
|
Lehnertz B, Pabst C, Su L, Miller M, Liu F, Yi L, Zhang R, Krosl J, Yung E, Kirschner J, Rosten P, Underhill TM, Jin J, Hébert J, Sauvageau G, Humphries RK, Rossi FM. The methyltransferase G9a regulates HoxA9-dependent transcription in AML. Genes Dev 2014; 28:317-27. [PMID: 24532712 PMCID: PMC3937511 DOI: 10.1101/gad.236794.113] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Lehnertz et al. identify the histone methyltransferase G9a as a selective regulator of fast proliferating myeloid progenitors with no discernible function in hematopoietic stem cells. Loss of G9a significantly delays disease progression and reduces leukemia stem cell frequency in mouse models of acute myeloid leukemia. G9a interacts with the leukemogenic transcription factor HoxA9 and regulates HoxA9-dependent transcription. These results highlight G9a inhibition as a means to counteract the proliferation and self-renewal of AML cells. Chromatin modulators are emerging as attractive drug targets, given their widespread implication in human cancers and susceptibility to pharmacological inhibition. Here we establish the histone methyltransferase G9a/EHMT2 as a selective regulator of fast proliferating myeloid progenitors with no discernible function in hematopoietic stem cells (HSCs). In mouse models of acute myeloid leukemia (AML), loss of G9a significantly delays disease progression and reduces leukemia stem cell (LSC) frequency. We connect this function of G9a to its methyltransferase activity and its interaction with the leukemogenic transcription factor HoxA9 and provide evidence that primary human AML cells are sensitive to G9A inhibition. Our results highlight a clinical potential of G9A inhibition as a means to counteract the proliferation and self-renewal of AML cells by attenuating HoxA9-dependent transcription.
Collapse
Affiliation(s)
- Bernhard Lehnertz
- University of British Columbia, Biomedical Research Centre, Vancouver, British Columbia V6T 1Z3, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
117
|
Martin N, Raguz S, Dharmalingam G, Gil J. Co-regulation of senescence-associated genes by oncogenic homeobox proteins and polycomb repressive complexes. Cell Cycle 2014; 12:2194-9. [PMID: 24067365 DOI: 10.4161/cc.25331] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Cellular senescence is a stable cell cycle arrest that can be induced by stresses such as telomere shortening, oncogene activation or DNA damage. Senescence is a potent anticancer barrier that needs to be circumvented during tumorigenesis. The cell cycle regulator p16(INK4a) is a key effector upregulated during senescence. Polycomb repressive complexes (PRCs) play a crucial role in silencing the INK4/ARF locus, which encodes for p16(INK4a), but the mechanisms by which PRCs are recruited to this locus as well as to other targets remain poorly understood. Recently we discovered the ability of the homeobox proteins HLX1 (H2.0-like homeobox 1) and HOXA9 (Homeobox A9) to bypass senescence. We showed that HLX1 and HOXA9 recruit PRCs to repress INK4a, which constitutes a key mechanism explaining their effects on senescence. Here we provide evidence for the regulation of additional senescence-associated PRC target genes by HLX1 and HOXA9. As both HLX1 and HOXA9 are oncogenes implicated in leukemogenesis, we discuss the implications that the collaboration between Homeobox proteins and PRCs has for senescence and cancer.
Collapse
Affiliation(s)
- Nadine Martin
- Cell Proliferation Group, MRC Clinical Sciences Centre, Imperial College London, London, UK
| | | | | | | |
Collapse
|
118
|
The leukemogenicity of Hoxa9 depends on alternative splicing. Leukemia 2014; 28:1838-43. [PMID: 24535405 DOI: 10.1038/leu.2014.74] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Revised: 02/03/2014] [Accepted: 02/07/2014] [Indexed: 01/22/2023]
Abstract
Although the transforming potential of Hox genes is known for a long time, it is not precisely understood to which extent splicing is important for the leukemogenicity of this gene family. To test this for Hoxa9, we compared the leukemogenic potential of the wild-type Hoxa9, which undergoes natural splicing, with a full-length Hoxa9 construct, which was engineered to prevent natural splicing (Hoxa9FLim). Inability to undergo splicing significantly reduced in vivo leukemogenicity compared to Hoxa9-wild-typed. Importantly, Hoxa9FLim could compensate for the reduced oncogenicity by collaborating with the natural splice variant Hoxa9T, as co-expression of Hoxa9T and Hoxa9FLim induced acute myeloid leukemia (AML) after a comparable latency time as wild-type Hoxa9. Hoxa9T on its own induced AML after a similar latency as Hoxa9FLim, despite its inability to bind DNA. These data assign splicing a central task in Hox gene mediated leukemogenesis and suggest an important role of homeodomain-less splice variants in hematological neoplasms.
Collapse
|
119
|
Gough SM, Lee F, Yang F, Walker RL, Zhu YJ, Pineda M, Onozawa M, Chung YJ, Bilke S, Wagner EK, Denu JM, Ning Y, Xu B, Wang GG, Meltzer PS, Aplan PD. NUP98-PHF23 is a chromatin-modifying oncoprotein that causes a wide array of leukemias sensitive to inhibition of PHD histone reader function. Cancer Discov 2014; 4:564-77. [PMID: 24535671 DOI: 10.1158/2159-8290.cd-13-0419] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
In this report, we show that expression of a NUP98-PHF23 (NP23) fusion, associated with acute myeloid leukemia (AML) in humans, leads to myeloid, erythroid, T-cell, and B-cell leukemia in mice. The leukemic and preleukemic tissues display a stem cell-like expression signature, including Hoxa, Hoxb, and Meis1 genes. The PHF23 plant homeodomain (PHD) motif is known to bind to H3K4me3 residues, and chromatin immunoprecipitation experiments demonstrated that the NP23 protein binds to chromatin at a specific subset of H3K4me3 sites, including at Hoxa, Hoxb, and Meis1. Treatment of NP23 cells with disulfiram, which inhibits the binding of PHD motifs to H3K4me3, rapidly and selectively killed NP23-expressing myeloblasts; cell death was preceded by decreased expression of Hoxa, Hoxb, and Meis1. Furthermore, AML driven by a related fusion gene, NUP98-JARID1A (NJL), was also sensitive to disulfiram. Thus, the NP23 mouse provides a platform to evaluate compounds that disrupt binding of oncogenic PHD proteins to H3K4me3.
Collapse
Affiliation(s)
- Sheryl M Gough
- 1Genetics Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda; 2Department of Pathology, Johns Hopkins University, Baltimore, Maryland; 3Department of Biomolecular Chemistry, University of Wisconsin, Madison, Wisconsin; and 4Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
120
|
Patel B, Kang Y, Cui K, Litt M, Riberio MSJ, Deng C, Salz T, Casada S, Fu X, Qiu Y, Zhao K, Huang S. Aberrant TAL1 activation is mediated by an interchromosomal interaction in human T-cell acute lymphoblastic leukemia. Leukemia 2014; 28:349-61. [PMID: 23698277 PMCID: PMC10921969 DOI: 10.1038/leu.2013.158] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 05/09/2013] [Accepted: 05/16/2013] [Indexed: 01/21/2023]
Abstract
Long-range chromatin interactions control metazoan gene transcription. However, the involvement of intra- and interchromosomal interactions in development and oncogenesis remains unclear. TAL1/SCL is a critical transcription factor required for the development of all hematopoietic lineages; yet, aberrant TAL1 transcription often occurs in T-cell acute lymphoblastic leukemia (T-ALL). Here, we report that oncogenic TAL1 expression is regulated by different intra- and interchromosomal loops in normal hematopoietic and leukemic cells, respectively. These intra- and interchromosomal loops alter the cell-type-specific enhancers that interact with the TAL1 promoter. We show that human SET1 (hSET1)-mediated H3K4 methylations promote a long-range chromatin loop, which brings the +51 enhancer in close proximity to TAL1 promoter 1 in erythroid cells. The CCCTC-binding factor (CTCF) facilitates this long-range enhancer/promoter interaction of the TAL1 locus in erythroid cells while blocking the same enhancer/promoter interaction of the TAL1 locus in human T-cell leukemia. In human T-ALL, a T-cell-specific transcription factor c-Maf-mediated interchromosomal interaction brings the TAL1 promoter into close proximity with a T-cell-specific regulatory element located on chromosome 16, activating aberrant TAL1 oncogene expression. Thus, our study reveals a novel molecular mechanism involving changes in three-dimensional chromatin interactions that activate the TAL1 oncogene in human T-cell leukemia.
Collapse
Affiliation(s)
- B Patel
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA
- These authors contributed equally to this work
| | - Y Kang
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA
- College of Life Science, Jilin University, Changchun, China
- These authors contributed equally to this work
| | - K Cui
- Center for System Biology, NHLBI, National Institute of Health, Bethesda, MD, USA
| | - M Litt
- Medical Education Center, Ball State University, Muncie, IN, USA
| | - MSJ Riberio
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - C Deng
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - T Salz
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - S Casada
- Medical Education Center, Ball State University, Muncie, IN, USA
| | - X Fu
- College of Life Science, Jilin University, Changchun, China
| | - Y Qiu
- Department of Anatomy and Cell Biology, College of Medicine, University of Florida, Gainesville, FL, USA
- Shands Cancer Center, College of Medicine, University of Florida, Gainesville, FL, USA
| | - K Zhao
- Center for System Biology, NHLBI, National Institute of Health, Bethesda, MD, USA
| | - S Huang
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA
- Shands Cancer Center, College of Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
121
|
Abstract
Roe and Vakoc discuss the role of C/EBPα in the initiation of acute myeloid leukemia. Acute myeloid leukemia (AML) is a hematopoietic malignancy characterized by clonal expansion of myeloid progenitor cells. A major mechanistic theme in AML biology is the extensive collaboration among fusion oncoproteins, transcription factors, and chromatin regulators to initiate and sustain a transformed cellular state. A new study in this issue describes how the C/EBPα transcription factor is crucial for the initiation of AML induced by MLL fusion oncoproteins, but is entirely dispensable for the maintenance of established disease. These observations provide a unique glimpse into the pioneer round of regulatory events that are critical at the origin of AML formation. Furthermore, this study implies the existence of oncogene-induced positive feedback loops capable of bypassing the continuous need for certain regulators to propagate disease.
Collapse
Affiliation(s)
- Jae-Seok Roe
- the Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | | |
Collapse
|
122
|
Velu CS, Chaubey A, Phelan JD, Horman SR, Wunderlich M, Guzman ML, Jegga AG, Zeleznik-Le NJ, Chen J, Mulloy JC, Cancelas JA, Jordan CT, Aronow BJ, Marcucci G, Bhat B, Gebelein B, Grimes HL. Therapeutic antagonists of microRNAs deplete leukemia-initiating cell activity. J Clin Invest 2014; 124:222-36. [PMID: 24334453 PMCID: PMC3871218 DOI: 10.1172/jci66005] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 10/10/2013] [Indexed: 12/14/2022] Open
Abstract
Acute myelogenous leukemia (AML) subtypes that result from oncogenic activation of homeobox (HOX) transcription factors are associated with poor prognosis. The HOXA9 transcription activator and growth factor independent 1 (GFI1) transcriptional repressor compete for occupancy at DNA-binding sites for the regulation of common target genes. We exploited this HOXA9 versus GFI1 antagonism to identify the genes encoding microRNA-21 and microRNA-196b as transcriptional targets of HOX-based leukemia oncoproteins. Therapeutic inhibition of microRNA-21 and microRNA-196b inhibited in vitro leukemic colony forming activity and depleted in vivo leukemia-initiating cell activity of HOX-based leukemias, which led to leukemia-free survival in a murine AML model and delayed disease onset in xenograft models. These data establish microRNA as functional effectors of endogenous HOXA9 and HOX-based leukemia oncoproteins, provide a concise in vivo platform to test RNA therapeutics, and suggest therapeutic value for microRNA antagonists in AML.
Collapse
MESH Headings
- Animals
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Base Sequence
- Binding Sites
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Combined Modality Therapy
- Cytarabine/administration & dosage
- DNA-Binding Proteins/metabolism
- Doxorubicin/administration & dosage
- Gene Expression Regulation, Leukemic
- Homeodomain Proteins/metabolism
- Humans
- Induction Chemotherapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/therapy
- Mice
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Mice, SCID
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Myeloid Ecotropic Viral Integration Site 1 Protein
- Neoplasm Proteins/metabolism
- Neoplastic Stem Cells/physiology
- Phosphorothioate Oligonucleotides/genetics
- Pre-B-Cell Leukemia Transcription Factor 1
- Protein Binding
- Proto-Oncogene Proteins/metabolism
- Regulatory Sequences, Nucleic Acid
- Transcription Factors/metabolism
- Transcriptome
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Chinavenmeni S. Velu
- Division of Immunobiology and
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Weill Cornell College of Medicine, New York, New York, USA.
Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Loyola University Medical Center, Maywood, Illinois, USA.
University of Chicago, Chicago, Illinois, USA.
University of Colorado, Aurora, Colorado, USA.
The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA.
Regulus Therapeutics, San Diego, California, USA.
Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Aditya Chaubey
- Division of Immunobiology and
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Weill Cornell College of Medicine, New York, New York, USA.
Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Loyola University Medical Center, Maywood, Illinois, USA.
University of Chicago, Chicago, Illinois, USA.
University of Colorado, Aurora, Colorado, USA.
The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA.
Regulus Therapeutics, San Diego, California, USA.
Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - James D. Phelan
- Division of Immunobiology and
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Weill Cornell College of Medicine, New York, New York, USA.
Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Loyola University Medical Center, Maywood, Illinois, USA.
University of Chicago, Chicago, Illinois, USA.
University of Colorado, Aurora, Colorado, USA.
The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA.
Regulus Therapeutics, San Diego, California, USA.
Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Shane R. Horman
- Division of Immunobiology and
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Weill Cornell College of Medicine, New York, New York, USA.
Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Loyola University Medical Center, Maywood, Illinois, USA.
University of Chicago, Chicago, Illinois, USA.
University of Colorado, Aurora, Colorado, USA.
The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA.
Regulus Therapeutics, San Diego, California, USA.
Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Mark Wunderlich
- Division of Immunobiology and
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Weill Cornell College of Medicine, New York, New York, USA.
Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Loyola University Medical Center, Maywood, Illinois, USA.
University of Chicago, Chicago, Illinois, USA.
University of Colorado, Aurora, Colorado, USA.
The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA.
Regulus Therapeutics, San Diego, California, USA.
Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Monica L. Guzman
- Division of Immunobiology and
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Weill Cornell College of Medicine, New York, New York, USA.
Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Loyola University Medical Center, Maywood, Illinois, USA.
University of Chicago, Chicago, Illinois, USA.
University of Colorado, Aurora, Colorado, USA.
The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA.
Regulus Therapeutics, San Diego, California, USA.
Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Anil G. Jegga
- Division of Immunobiology and
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Weill Cornell College of Medicine, New York, New York, USA.
Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Loyola University Medical Center, Maywood, Illinois, USA.
University of Chicago, Chicago, Illinois, USA.
University of Colorado, Aurora, Colorado, USA.
The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA.
Regulus Therapeutics, San Diego, California, USA.
Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Nancy J. Zeleznik-Le
- Division of Immunobiology and
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Weill Cornell College of Medicine, New York, New York, USA.
Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Loyola University Medical Center, Maywood, Illinois, USA.
University of Chicago, Chicago, Illinois, USA.
University of Colorado, Aurora, Colorado, USA.
The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA.
Regulus Therapeutics, San Diego, California, USA.
Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Jianjun Chen
- Division of Immunobiology and
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Weill Cornell College of Medicine, New York, New York, USA.
Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Loyola University Medical Center, Maywood, Illinois, USA.
University of Chicago, Chicago, Illinois, USA.
University of Colorado, Aurora, Colorado, USA.
The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA.
Regulus Therapeutics, San Diego, California, USA.
Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - James C. Mulloy
- Division of Immunobiology and
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Weill Cornell College of Medicine, New York, New York, USA.
Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Loyola University Medical Center, Maywood, Illinois, USA.
University of Chicago, Chicago, Illinois, USA.
University of Colorado, Aurora, Colorado, USA.
The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA.
Regulus Therapeutics, San Diego, California, USA.
Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Jose A. Cancelas
- Division of Immunobiology and
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Weill Cornell College of Medicine, New York, New York, USA.
Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Loyola University Medical Center, Maywood, Illinois, USA.
University of Chicago, Chicago, Illinois, USA.
University of Colorado, Aurora, Colorado, USA.
The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA.
Regulus Therapeutics, San Diego, California, USA.
Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Craig T. Jordan
- Division of Immunobiology and
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Weill Cornell College of Medicine, New York, New York, USA.
Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Loyola University Medical Center, Maywood, Illinois, USA.
University of Chicago, Chicago, Illinois, USA.
University of Colorado, Aurora, Colorado, USA.
The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA.
Regulus Therapeutics, San Diego, California, USA.
Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Bruce J. Aronow
- Division of Immunobiology and
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Weill Cornell College of Medicine, New York, New York, USA.
Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Loyola University Medical Center, Maywood, Illinois, USA.
University of Chicago, Chicago, Illinois, USA.
University of Colorado, Aurora, Colorado, USA.
The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA.
Regulus Therapeutics, San Diego, California, USA.
Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Guido Marcucci
- Division of Immunobiology and
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Weill Cornell College of Medicine, New York, New York, USA.
Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Loyola University Medical Center, Maywood, Illinois, USA.
University of Chicago, Chicago, Illinois, USA.
University of Colorado, Aurora, Colorado, USA.
The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA.
Regulus Therapeutics, San Diego, California, USA.
Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Balkrishen Bhat
- Division of Immunobiology and
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Weill Cornell College of Medicine, New York, New York, USA.
Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Loyola University Medical Center, Maywood, Illinois, USA.
University of Chicago, Chicago, Illinois, USA.
University of Colorado, Aurora, Colorado, USA.
The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA.
Regulus Therapeutics, San Diego, California, USA.
Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Brian Gebelein
- Division of Immunobiology and
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Weill Cornell College of Medicine, New York, New York, USA.
Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Loyola University Medical Center, Maywood, Illinois, USA.
University of Chicago, Chicago, Illinois, USA.
University of Colorado, Aurora, Colorado, USA.
The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA.
Regulus Therapeutics, San Diego, California, USA.
Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - H. Leighton Grimes
- Division of Immunobiology and
Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Weill Cornell College of Medicine, New York, New York, USA.
Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA.
Loyola University Medical Center, Maywood, Illinois, USA.
University of Chicago, Chicago, Illinois, USA.
University of Colorado, Aurora, Colorado, USA.
The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA.
Regulus Therapeutics, San Diego, California, USA.
Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
123
|
PU.1 is essential for MLL leukemia partially via crosstalk with the MEIS/HOX pathway. Leukemia 2013; 28:1436-48. [PMID: 24445817 DOI: 10.1038/leu.2013.384] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 11/22/2013] [Accepted: 12/12/2013] [Indexed: 01/07/2023]
Abstract
Mixed lineage leukemia (MLL) fusion proteins directly activate the expression of key downstream genes such as MEIS1, HOXA9 to drive an aggressive form of human leukemia. However, it is still poorly understood what additional transcriptional regulators, independent of the MLL fusion pathway, contribute to the development of MLL leukemia. Here we show that the transcription factor PU.1 is essential for MLL leukemia and is required for the growth of MLL leukemic cells via the promotion of cell-cycle progression and inhibition of apoptosis. Importantly, PU.1 expression is not under the control of MLL fusion proteins. We further identified a PU.1-governed 15-gene signature, which contains key regulators in the MEIS-HOX program (MEIS1, PBX3, FLT3, and c-KIT). PU.1 directly binds to the genomic loci of its target genes in vivo, and is required to maintain active expression of those genes in both normal hematopoietic stem and progenitor cells and in MLL leukemia. Finally, the clinical significance of the identified PU.1 signature was indicated by its ability to predict survival in acute myelogenous leukemia patients. Together, our findings demonstrate that PU.1 contributes to the development of MLL leukemia, partially via crosstalk with the MEIS/HOX pathway.
Collapse
|
124
|
|
125
|
A Cdx4-Sall4 regulatory module controls the transition from mesoderm formation to embryonic hematopoiesis. Stem Cell Reports 2013; 1:425-36. [PMID: 24286030 PMCID: PMC3841246 DOI: 10.1016/j.stemcr.2013.10.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Revised: 10/01/2013] [Accepted: 10/02/2013] [Indexed: 12/26/2022] Open
Abstract
Deletion of caudal/cdx genes alters hox gene expression and causes defects in posterior tissues and hematopoiesis. Yet, the defects in hox gene expression only partially explain these phenotypes. To gain deeper insight into Cdx4 function, we performed chromatin immunoprecipitation sequencing (ChIP-seq) combined with gene-expression profiling in zebrafish, and identified the transcription factor spalt-like 4 (sall4) as a Cdx4 target. ChIP-seq revealed that Sall4 bound to its own gene locus and the cdx4 locus. Expression profiling showed that Cdx4 and Sall4 coregulate genes that initiate hematopoiesis, such as hox, scl, and lmo2. Combined cdx4/sall4 gene knockdown impaired erythropoiesis, and overexpression of the Cdx4 and Sall4 target genes scl and lmo2 together rescued the erythroid program. These findings suggest that auto- and cross-regulation of Cdx4 and Sall4 establish a stable molecular circuit in the mesoderm that facilitates the activation of the blood-specific program as development proceeds. Cdx4 and Sall4 bind to each other’s genomic loci Cdx4 and Sall4 coregulate genes responsible for the mesoderm-to-blood transition Scl and Lmo2 overexpression rescues blood defects in cdx4/sall4 double morphants
Collapse
|
126
|
Peptide-based inhibition of the HOXA9/PBX interaction retards the growth of human meningioma. Cancer Chemother Pharmacol 2013; 73:53-60. [PMID: 24141373 DOI: 10.1007/s00280-013-2316-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2013] [Accepted: 10/04/2013] [Indexed: 01/22/2023]
Abstract
BACKGROUND Meningiomas are the most common type of intracranial tumor, accounting for between 24 and 30 % of primary intracranial tumors. Thus far, no biomarkers exist to reliably predict the clinical outcome of meningiomas. A previous genome-wide methylation analysis revealed that HOXA9 is one of the most functionally relevant biomarkers. In this study, we have examined whether HOXA9 is a potential therapeutic target in meningiomas, using HXR9, a peptide inhibitor of the interaction between HOXA9 and its cofactor PBX. METHODS We determined the expression level of HOXA9 in human meningiomas, meningioma cell lines, and normal brain tissue. Meningioma in culture and in subcutaneous tumors was treated with HXR9. We also examined the disruption of HOXA9/PBX dimers. RESULTS We first confirmed that HOXA9 is highly expressed in meningiomas, but not in normal brain tissue. The HXR9 peptide blocks the binding of HOXA9 to PBX, leading to an alteration of DNA binding, and subsequent regulation of their target genes. HXR9 markedly inhibited the growth of meningioma cells and subcutaneous meningeal tumors. CONCLUSION There is no effective chemotherapy for meningiomas at present, and targeting the HOXA9/PBX interaction may represent a novel treatment option for this disease.
Collapse
|
127
|
Hassawi M, Shestakova EA, Fournier M, Lebert-Ghali CÉ, Vaisson G, Frison H, Sinnett D, Vidal R, Thompson A, Bijl JJ. Hoxa9 collaborates with E2A-PBX1 in mouse B cell leukemia in association with Flt3 activation and decrease of B cell gene expression. Dev Dyn 2013; 243:145-58. [PMID: 23996689 DOI: 10.1002/dvdy.24056] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Revised: 08/26/2013] [Accepted: 08/27/2013] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The fusion protein E2A-PBX1 induces pediatric B cell leukemia in human. Previously, we reported oncogenic interactions between homeobox (Hox) genes and E2A-PBX1 in murine T cell leukemia. A proviral insertional mutagenesis screen with our E2A-PBX1 B cell leukemia mouse model identified Hoxa genes as potential collaborators to E2A-PBX1. Here we studied whether Hoxa9 could enhance E2A-PBX1 leukemogenesis. RESULTS We show that Hoxa9 confers a proliferative advantage to E2A-PBX1 B cells. Transplantation experiments with E2A-PBX1 transgenic B cells overexpressing Hoxa9 isolated from bone marrow chimeras showed that Hoxa9 accelerates the generation of E2A-PBX1 B cell leukemia, but Hoxa9 is unable to transform B cells alone. Quantitative-reverse transcriptase polymerase chain reaction analysis demonstrated a strong repression of B cell specific genes in these E2A-PBX1/Hoxa9 leukemias in addition to Flt3 activation, indicating inhibition of B cell differentiation in combination with enhanced proliferation. Overexpression of Hoxa9 in established E2A-PBX1 mouse leukemic B cells resulted in a growth advantage in vitro, which was also characterized by an enhanced expression of Flt3. CONCLUSIONS we show for the first time that Hoxa9 collaborates with E2A-PBX1 in the oncogenic transformation of B cells in a mouse model that involves Flt3 signaling, which is potentially relevant to human disease.
Collapse
Affiliation(s)
- Mona Hassawi
- Hospital Maisonneuve-Rosemont Research Center, Montreal, QC
| | | | | | | | | | | | | | | | | | | |
Collapse
|
128
|
Doulatov S, Vo LT, Chou SS, Kim PG, Arora N, Li H, Hadland BK, Bernstein ID, Collins JJ, Zon LI, Daley GQ. Induction of multipotential hematopoietic progenitors from human pluripotent stem cells via respecification of lineage-restricted precursors. Cell Stem Cell 2013; 13:459-70. [PMID: 24094326 PMCID: PMC3888026 DOI: 10.1016/j.stem.2013.09.002] [Citation(s) in RCA: 222] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Revised: 08/20/2013] [Accepted: 09/06/2013] [Indexed: 01/19/2023]
Abstract
Human pluripotent stem cells (hPSCs) represent a promising source of patient-specific cells for disease modeling, drug screens, and cellular therapies. However, the inability to derive engraftable human hematopoietic stem and progenitor cells (HSPCs) has limited their characterization to in vitro assays. We report a strategy to respecify lineage-restricted CD34(+)CD45(+) myeloid precursors derived from hPSCs into multilineage progenitors that can be expanded in vitro and engrafted in vivo. HOXA9, ERG, and RORA conferred self-renewal and multilineage potential in vitro and maintained primitive CD34(+)CD38(-) cells. Screening cells via transplantation revealed that two additional factors, SOX4 and MYB, conferred engraftment. Progenitors specified with all five factors gave rise to reproducible short-term engraftment with myeloid and erythroid lineages. Erythroid precursors underwent hemoglobin switching in vivo, silencing embryonic and activating adult globin expression. Our combinatorial screening approach establishes a strategy for obtaining transcription-factor-mediated engraftment of blood progenitors from human pluripotent cells.
Collapse
Affiliation(s)
- Sergei Doulatov
- Stem Cell Transplantation Program, Division of Pediatric Hematology/Oncology, Manton Center for Orphan Disease Research, Howard Hughes Medical Institute, Children's Hospital Boston and Dana Farber Cancer Institute, Boston, MA, 02115
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115
| | - Linda T. Vo
- Stem Cell Transplantation Program, Division of Pediatric Hematology/Oncology, Manton Center for Orphan Disease Research, Howard Hughes Medical Institute, Children's Hospital Boston and Dana Farber Cancer Institute, Boston, MA, 02115
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115
| | - Stephanie S. Chou
- Stem Cell Transplantation Program, Division of Pediatric Hematology/Oncology, Manton Center for Orphan Disease Research, Howard Hughes Medical Institute, Children's Hospital Boston and Dana Farber Cancer Institute, Boston, MA, 02115
| | - Peter G. Kim
- Stem Cell Transplantation Program, Division of Pediatric Hematology/Oncology, Manton Center for Orphan Disease Research, Howard Hughes Medical Institute, Children's Hospital Boston and Dana Farber Cancer Institute, Boston, MA, 02115
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115
| | - Natasha Arora
- Stem Cell Transplantation Program, Division of Pediatric Hematology/Oncology, Manton Center for Orphan Disease Research, Howard Hughes Medical Institute, Children's Hospital Boston and Dana Farber Cancer Institute, Boston, MA, 02115
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115
| | - Hu Li
- Howard Hughes Medical Institute, Department of Biomedical Engineering and Center for BioDynamics, Boston University, Boston, MA, 02215
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115
| | - Brandon K. Hadland
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109
- Department of Pediatrics, University of Washington, Seattle, WA, 98105
| | - Irwin D. Bernstein
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109
- Department of Pediatrics, University of Washington, Seattle, WA, 98105
| | - James J. Collins
- Howard Hughes Medical Institute, Department of Biomedical Engineering and Center for BioDynamics, Boston University, Boston, MA, 02215
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115
| | - Leonard I. Zon
- Stem Cell Transplantation Program, Division of Pediatric Hematology/Oncology, Manton Center for Orphan Disease Research, Howard Hughes Medical Institute, Children's Hospital Boston and Dana Farber Cancer Institute, Boston, MA, 02115
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115
- Harvard Stem Cell Institute, Boston, MA, 02138
- Broad Institute, Boston, MA, 02141
| | - George Q. Daley
- Stem Cell Transplantation Program, Division of Pediatric Hematology/Oncology, Manton Center for Orphan Disease Research, Howard Hughes Medical Institute, Children's Hospital Boston and Dana Farber Cancer Institute, Boston, MA, 02115
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115
- Harvard Stem Cell Institute, Boston, MA, 02138
- Broad Institute, Boston, MA, 02141
| |
Collapse
|
129
|
Regulation of MEIS1 by distal enhancer elements in acute leukemia. Leukemia 2013; 28:138-46. [PMID: 24022755 PMCID: PMC5774621 DOI: 10.1038/leu.2013.260] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 08/22/2013] [Accepted: 08/29/2013] [Indexed: 11/15/2022]
Abstract
Aberrant activation of the three-amino-acid-loop extension (TALE) homeobox gene MEIS1 shortens the latency and accelerates the onset and progression of acute leukemia, yet the molecular mechanism underlying persistent activation of the MEIS1 gene in leukemia remains poorly understood. Here we used a combined comparative genomics analysis and an in vivo transgenic zebrafish assay to identify 6 regulatory DNA elements that are able to direct GFP expression in a spatiotemporal manner during zebrafish embryonic hematopoiesis. Analysis of chromatin characteristics and regulatory signatures suggest that many of these predicted elements are potential enhancers in mammalian hematopoiesis. Strikingly, one of the enhancer elements (E9) is a frequent integration site in retroviral induced mouse acute leukemia. The genomic region corresponding to enhancer E9 is differentially marked by H3K4 mono-methylation and H3K27 acetylation, hallmarks of active enhancers, in multiple leukemia cell lines. Decreased enrichment of these histone marks is associated with downregulation of MEIS1 expression during hematopoietic differentiation. Furthermore, MEIS1/HOXA9 transactivate this enhancer via a conserved binding motif in vitro, and participate in an autoregulatory loop that modulates MEIS1 expression in vivo. Our results suggest that an intronic enhancer regulates the expression of MEIS1 in hematopoiesis and contributes to its aberrant expression in acute leukemia.
Collapse
|
130
|
Transcriptional profiling of mRNAs and microRNAs in human bone marrow precursor B cells identifies subset- and age-specific variations. PLoS One 2013; 8:e70721. [PMID: 23936243 PMCID: PMC3728296 DOI: 10.1371/journal.pone.0070721] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 06/20/2013] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Molecular mechanisms explaining age-related changes in the bone marrow with reduced precursor B cell output are poorly understood. METHODS We studied the transcriptome of five precursor B cell subsets in individual bone marrow samples from 4 healthy children and 4 adults employing GeneChip® Human Exon 1.0 ST Arrays (Affymetrix®) and TaqMan® Array MicroRNA Cards (Life Technologies™). RESULTS A total of 1796 mRNAs (11%) were at least once differentially expressed between the various precursor B cell subsets in either age group (FDR 0.1%, p≤1.13×10(-4)) with more marked cell stage specific differences than those related to age. In contrast, microRNA profiles of the various precursor B cell subsets showed less hierarchical clustering as compared to the corresponding mRNA profiles. However, 17 of the 667 microRNA assays (2.5%) were at least once differentially expressed between the subsets (FDR 10%, p≤0.004). From target analysis (Ingenuity® Systems), functional assignment between postulated interacting mRNAs and microRNAs showed especially association to cellular growth, proliferation and cell cycle regulation. One functional network connected up-regulation of the differentiation inhibitor ID2 mRNA to down-regulation of the hematopoiesis- or cell cycle regulating miR-125b-5p, miR-181a-5p, miR-196a-5p, miR-24-3p and miR-320d in adult PreBII large cells. Noteworthy was also the stage-dependent expression of the growth promoting miR-17-92 cluster, showing a partly inverse trend with age, reaching statistical significance at the PreBII small stage (up 3.1-12.9 fold in children, p = 0.0084-0.0270). CONCLUSIONS The global mRNA profile is characteristic for each precursor B cell developmental stage and largely similar in children and adults. The microRNA profile is much cell stage specific and not changing much with age. Importantly, however, specific age-dependent differences involving key networks like differentiation and cellular growth may indicate biological divergence and possibly also altered production potential with age.
Collapse
|
131
|
Ramsey JM, Kettyle LM, Sharpe DJ, Mulgrew NM, Dickson GJ, Bijl JJ, Austin P, Mayotte N, Cellot S, Lappin TR, Zhang SD, Mills KI, Krosl J, Sauvageau G, Thompson A. Entinostat Prevents Leukemia Maintenance in a Collaborating Oncogene-Dependent Model of Cytogenetically Normal Acute Myeloid Leukemia. Stem Cells 2013; 31:1434-45. [DOI: 10.1002/stem.1398] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2012] [Accepted: 03/14/2013] [Indexed: 12/16/2022]
|
132
|
Volpe G, Walton DS, Del Pozzo W, Garcia P, Dassé E, O’Neill LP, Griffiths M, Frampton J, Dumon S. C/EBPα and MYB regulate FLT3 expression in AML. Leukemia 2013; 27:1487-96. [PMID: 23340802 PMCID: PMC4214120 DOI: 10.1038/leu.2013.23] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Revised: 12/22/2012] [Accepted: 01/09/2013] [Indexed: 11/09/2022]
Abstract
The interaction between the receptor FLT3 (FMS-like tyrosine kinase-3) and its ligand FL leads to crucial signalling during the early stages of the commitment of haematopoietic stem cells. Mutation or over-expression of the FLT3 gene, leading to constitutive signalling, enhances the survival and expansion of a variety of leukaemias and is associated with an unfavourable clinical outcome for acute myeloid leukaemia (AML) patients. In this study, we used a murine cellular model for AML and primary leukaemic cells from AML patients to investigate the molecular mechanisms underlying the regulation of FLT3 gene expression and identify its key cis- and trans-regulators. By assessing DNA accessibility and epigenetic markings, we defined regulatory domains in the FLT3 promoter and first intron. These elements permit in vivo binding of several AML-related transcription factors, including the proto-oncogene MYB and the CCAAT/enhancer binding protein C/EBPα, which are recruited to the FLT3 promoter and intronic module, respectively. Substantiating their relevance to the human disease, our analysis of gene expression profiling arrays from AML patients uncovered significant correlations between FLT3 expression level and that of MYB and CEBPA. The latter relationship permits discrimination between patients with CEBPA mono- and bi-allelic mutations, and thus connects two major prognostic factors for AML.
Collapse
MESH Headings
- Animals
- Base Sequence
- CCAAT-Enhancer-Binding Proteins/genetics
- CCAAT-Enhancer-Binding Proteins/metabolism
- Cell Line, Tumor
- Disease Models, Animal
- Epigenesis, Genetic/physiology
- Gene Expression Regulation, Leukemic/physiology
- Genetic Complementation Test
- Homeodomain Proteins/genetics
- Humans
- Introns/genetics
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Mice
- Molecular Sequence Data
- Myeloid Ecotropic Viral Integration Site 1 Protein
- Neoplasm Proteins/genetics
- Promoter Regions, Genetic/genetics
- Proto-Oncogene Mas
- Proto-Oncogene Proteins c-myb/genetics
- Proto-Oncogene Proteins c-myb/metabolism
- Tumor Cells, Cultured
- fms-Like Tyrosine Kinase 3/genetics
- fms-Like Tyrosine Kinase 3/metabolism
Collapse
Affiliation(s)
- G Volpe
- Institute of Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - DS Walton
- Institute of Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - W Del Pozzo
- Nikhef, Science Park, Amsterdam, XG, The Netherlands
| | - P Garcia
- Institute of Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - E Dassé
- Institute of Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - LP O’Neill
- Institute of Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - M Griffiths
- West Midlands Regional Genetics Laboratory, Birmingham Women’s NHS Foundation Trust, Birmingham, UK
| | - J Frampton
- Institute of Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - S Dumon
- Institute of Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
133
|
Abstract
The ten-eleven translocation 1 (TET1) gene is the founding member of the TET family of enzymes (TET1/2/3) that convert 5-methylcytosine to 5-hydroxymethylcytosine. Although TET1 was first identified as a fusion partner of the mixed lineage leukemia (MLL) gene in acute myeloid leukemia carrying t(10,11), its definitive role in leukemia is unclear. In contrast to the frequent down-regulation (or loss-of-function mutations) and critical tumor-suppressor roles of the three TET genes observed in various types of cancers, here we show that TET1 is a direct target of MLL-fusion proteins and is significantly up-regulated in MLL-rearranged leukemia, leading to a global increase of 5-hydroxymethylcytosine level. Furthermore, our both in vitro and in vivo functional studies demonstrate that Tet1 plays an indispensable oncogenic role in the development of MLL-rearranged leukemia, through coordination with MLL-fusion proteins in regulating their critical cotargets, including homeobox A9 (Hoxa9)/myeloid ecotropic viral integration 1 (Meis1)/pre-B-cell leukemia homeobox 3 (Pbx3) genes. Collectively, our data delineate an MLL-fusion/Tet1/Hoxa9/Meis1/Pbx3 signaling axis in MLL-rearranged leukemia and highlight TET1 as a potential therapeutic target in treating this presently therapy-resistant disease.
Collapse
|
134
|
Gwin KA, Shapiro MB, Dolence JJ, Huang ZL, Medina KL. Hoxa9 and Flt3 signaling synergistically regulate an early checkpoint in lymphopoiesis. THE JOURNAL OF IMMUNOLOGY 2013; 191:745-54. [PMID: 23772038 DOI: 10.4049/jimmunol.1203294] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Hoxa9 and Flt3 signaling are individually important for the generation of lymphoid lineage precursors from multipotent hematopoietic progenitors (MPP) in bone marrow. Mice deficient for Hoxa9, Flt3, or Flt3 ligand (FL) have reduced numbers of lymphoid-primed multipotential progenitors (LMPP), common lymphoid progenitors (CLP), and B/T cell precursors. Hoxa9 regulates lymphoid development, in part, through transcriptional regulation of Flt3. However, it was unclear whether Hoxa9 has functions in lymphopoiesis independent of, or alternatively, synergistically with Flt3 signaling. In this study, we show that Hoxa9(-/-)Flt3l(-/-) mice have more severe deficiencies in all B lineage cells, CLP, LMPP, and total Flt3(+) MPP in bone marrow than the single knockouts. Although LMPP and Flt3(+) CLP contain precursors for NK and dendritic cell lineage cells, no deficiencies in these lineages beyond that in Flt3l(-/-) mice was found. Thymocyte cellularity was significantly reduced in the compound knockout, although peripheral T cell numbers mirrored Flt3l(-/-) mice. Analysis of the hematopoietic progenitor compartment revealed elevated numbers of CD150(+hi)CD34(-)CD41(+) myeloid-biased stem cells in Hoxa9(-/-)Flt3l(-/-) mice. In contrast, CD150(-) MPP enriched for lymphoid potential were synergistically reduced, suggesting Hoxa9 and Flt3 signaling function coordinately to regulate lymphopoiesis at a very early stage. Real-time PCR analysis of CD150(-)Flt3(+) cells from wild-type control, Hoxa9(-/-), and Flt3l(-/-) single knockouts revealed decreased lymphoid transcripts, corroborating the importance of these regulators in lymphoid development. Taken together, these studies reveal a very early checkpoint in lymphopoiesis dependent on the combinatorial activities of Hoxa9 function and Flt3 signaling.
Collapse
Affiliation(s)
- Kimberly A Gwin
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | | | | |
Collapse
|
135
|
Foundational concepts of cell fate conversion to the hematopoietic lineage. Curr Opin Genet Dev 2013; 23:585-90. [PMID: 23725798 DOI: 10.1016/j.gde.2013.04.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Accepted: 04/05/2013] [Indexed: 12/20/2022]
Abstract
Direct conversion of cellular fate provides a potential approach to generate cells of the hematopoietic lineage without the requisite reversion to a pluripotent state via somatic cell reprogramming. The utilization of this technology has enabled transcription factor-mediated conversion of somatic cell types to primitive and mature hematopoietic cells. Recent studies demonstrate that the direct conversion of somatic cells to the hematopoietic lineage likely requires the use of pioneer transcription factors to establish an accessible chromatin state that is responsive to enforced expression of hematopoietic-specific transcription factors, in combination with appropriate culture conditions that facilitate reprogramming. Developing adaptable, experimental strategies that incorporate these parameters should enable the efficient generation of human hematopoietic cells with translational potential.
Collapse
|
136
|
Abstract
Advances in our understanding of the genetic determinants of leukemia have translated to better treatment options and improved survival of patients with acute myeloid and acute lymphoid leukemia. However, some leukemias, such as those bearing 11q23 (MLL) translocations, result in aggressive diseases with a relatively poor prognosis, despite improved treatments such as allogeneic hematopoietic stem cell transplantation. This article will briefly review the functions and regulation of wild-type MLL during normal hematopoiesis, while focusing on recent advances in our understanding of the molecular mechanisms governing MLL leukemias. The transcriptional targets, cooperating signaling pathways and molecular machinery involved in MLL-associated leukemias will be discussed, as well as how these may be harnessed for more personalized treatment of this disease.
Collapse
Affiliation(s)
- Andrew G Muntean
- Department of Pathology, Department of Medicine, University of Michigan Medical School, 7520B Medical Science Research Building I, 1301 Catherine Road, Ann Arbor, MI 48109-5602, USA
| |
Collapse
|
137
|
Calero-Nieto FJ, Joshi A, Bonadies N, Kinston S, Chan WI, Gudgin E, Pridans C, Landry JR, Kikuchi J, Huntly BJ, Gottgens B. HOX-mediated LMO2 expression in embryonic mesoderm is recapitulated in acute leukaemias. Oncogene 2013; 32:5471-80. [PMID: 23708655 PMCID: PMC3898495 DOI: 10.1038/onc.2013.175] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 02/25/2013] [Accepted: 03/31/2013] [Indexed: 01/02/2023]
Abstract
The Lim Domain Only 2 (LMO2) leukaemia oncogene encodes an LIM domain transcriptional cofactor required for early haematopoiesis. During embryogenesis, LMO2 is also expressed in developing tail and limb buds, an expression pattern we now show to be recapitulated in transgenic mice by an enhancer in LMO2 intron 4. Limb bud expression depended on a cluster of HOX binding sites, while posterior tail expression required the HOX sites and two E-boxes. Given the importance of both LMO2 and HOX genes in acute leukaemias, we further demonstrated that the regulatory hierarchy of HOX control of LMO2 is activated in leukaemia mouse models as well as in patient samples. Moreover, Lmo2 knock-down impaired the growth of leukaemic cells, and high LMO2 expression at diagnosis correlated with poor survival in cytogenetically normal AML patients. Taken together, these results establish a regulatory hierarchy of HOX control of LMO2 in normal development, which can be resurrected during leukaemia development. Redeployment of embryonic regulatory hierarchies in an aberrant context is likely to be relevant in human pathologies beyond the specific example of ectopic activation of LMO2.
Collapse
Affiliation(s)
- F J Calero-Nieto
- Department of Haematology, Wellcome Trust and MRC Cambridge Stem Cell Institute, Cambridge Institute for Medical Research, Cambridge University, Cambridge, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
138
|
Tchkonia T, Thomou T, Zhu Y, Karagiannides I, Pothoulakis C, Jensen MD, Kirkland JL. Mechanisms and metabolic implications of regional differences among fat depots. Cell Metab 2013; 17:644-656. [PMID: 23583168 PMCID: PMC3942783 DOI: 10.1016/j.cmet.2013.03.008] [Citation(s) in RCA: 492] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Fat distribution is closely linked to metabolic disease risk. Distribution varies with sex, genetic background, disease state, certain drugs and hormones, development, and aging. Preadipocyte replication and differentiation, developmental gene expression, susceptibility to apoptosis and cellular senescence, vascularity, inflammatory cell infiltration, and adipokine secretion vary among depots, as do fatty-acid handling and mechanisms of enlargement with positive-energy and loss with negative-energy balance. How interdepot differences in these molecular, cellular, and pathophysiological properties are related is incompletely understood. Whether fat redistribution causes metabolic disease or whether it is a marker of underlying processes that are primarily responsible is an open question.
Collapse
Affiliation(s)
| | - Thomas Thomou
- Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Yi Zhu
- Robert and Arlene Kogod Center on Aging
| | - Iordanes Karagiannides
- Inflammatory Bowel Disease Center, Division of Digestive Diseases, Department of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Charalabos Pothoulakis
- Inflammatory Bowel Disease Center, Division of Digestive Diseases, Department of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | | | | |
Collapse
|
139
|
Li Z, Zhang Z, Li Y, Arnovitz S, Chen P, Huang H, Jiang X, Hong GM, Kunjamma RB, Ren H, He C, Wang CZ, Elkahloun AG, Valk PJM, Döhner K, Neilly MB, Bullinger L, Delwel R, Löwenberg B, Liu PP, Morgan R, Rowley JD, Yuan CS, Chen J. PBX3 is an important cofactor of HOXA9 in leukemogenesis. Blood 2013; 121:1422-31. [PMID: 23264595 PMCID: PMC3578957 DOI: 10.1182/blood-2012-07-442004] [Citation(s) in RCA: 111] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Accepted: 12/02/2012] [Indexed: 01/07/2023] Open
Abstract
Although PBX proteins are known to increase DNA-binding/transcriptional activity of HOX proteins through their direct binding, the functional importance of their interaction in leukemogenesis is unclear.We recently reported that overexpression of a 4-homeobox-gene signature (ie, PBX3/HOXA7/HOXA9/HOXA11) is an independent predictor of poor survival in patients with cytogenetically abnormal acute myeloid leukemia (CA-AML). Here we show that it is PBX3, but not PBX1 or PBX2, that is consistently coexpressed with HOXA9 in various subtypes of CA-AML, particularly MLL-rearranged AML, and thus appears as a potential pathologic cofactor of HOXA9 in CA-AML. We then show that depletion of endogenous Pbx3 expression by shRNA significantly inhibits MLL-fusion-mediated cell transformation, and coexpressed PBX3 exhibits a significantly synergistic effect with HOXA9 in promoting cell transformation in vitro and leukemogenesis in vivo. Furthermore, as a proof of concept, we show that a small peptide, namely HXR9, which was developed to specifically disrupt the interactions between HOX and PBX proteins, can selectively kill leukemic cells with overexpression of HOXA/PBX3 genes. Collectively, our data suggest that PBX3 is a critical cofactor of HOXA9 in leukemogenesis, and targeting their interaction is a feasible strategy to treat presently therapy resistant CA-AML (eg, MLL-rearranged leukemia) in which HOXA/PBX3 genes are overexpressed.
Collapse
MESH Headings
- Animals
- Bone Marrow Cells/physiology
- Bone Marrow Transplantation
- Cell Line, Transformed
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Gene Expression Regulation, Leukemic/drug effects
- Gene Expression Regulation, Leukemic/physiology
- Gene Rearrangement/genetics
- HEK293 Cells
- Histone-Lysine N-Methyltransferase
- Homeodomain Proteins/antagonists & inhibitors
- Homeodomain Proteins/genetics
- Homeodomain Proteins/metabolism
- Humans
- Intercellular Signaling Peptides and Proteins
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Mutant Strains
- Myeloid-Lymphoid Leukemia Protein/genetics
- Peptides/pharmacology
- Pre-B-Cell Leukemia Transcription Factor 1
- Proto-Oncogene Proteins/antagonists & inhibitors
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/metabolism
- RNA, Small Interfering/genetics
- Rats
- Transcription Factors/genetics
- Transcription Factors/metabolism
Collapse
Affiliation(s)
- Zejuan Li
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL 60637, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
140
|
Gwin K, Dolence JJ, Shapiro MB, Medina KL. Differential requirement for Hoxa9 in the development and differentiation of B, NK, and DC-lineage cells from Flt3+ multipotential progenitors. BMC Immunol 2013; 14:5. [PMID: 23363389 PMCID: PMC3565875 DOI: 10.1186/1471-2172-14-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Accepted: 01/25/2013] [Indexed: 12/13/2022] Open
Abstract
Hoxa9 is a homeodomain transcription factor important for the generation of Flt3+hiIL-7R- lymphoid biased-multipotential progenitors, Flt3+IL-7R+ common lymphoid progenitors (CLPs), and B cell precursors (BCP) in bone marrow (BM). In addition to B-cell, Flt3+IL-7R+ CLPs possess NK and DC developmental potentials, although DCs arise from Flt3+IL-7R- myeloid progenitors as well. In this study, we investigated the requirement for Hoxa9, from Flt3+ or Flt3- progenitor subsets, in the development of NK and DC lineage cells in BM. Flt3+IL-7R+Ly6D- CLPs and their Flt3+IL-7R+Ly6D+ B lineage-restricted progeny (BLP) were significantly reduced in hoxa9−/− mice. Interestingly, the reduction in Flt3+IL-7R+ CLPs in hoxa9−/− mice had no impact on the generation of NK precursor (NKP) subsets, the differentiation of NKP into mature NK cells, or NK homeostasis. Similarly, percentages and numbers of common dendritic progenitors (CDP), as well as their plasmacytoid or conventional dendritic cell progeny in hoxa9−/− mice were comparable to wildtype. These findings reveal distinct requirements for Hoxa9 or Hoxa9/Flt3 molecular circuits in regulation of B versus NK and DC development in BM.
Collapse
Affiliation(s)
- Kimberly Gwin
- Department of Immunology, College of Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | | | | | | |
Collapse
|
141
|
Chowdhury AH, Ramroop JR, Upadhyay G, Sengupta A, Andrzejczyk A, Saleque S. Differential transcriptional regulation of meis1 by Gfi1b and its co-factors LSD1 and CoREST. PLoS One 2013; 8:e53666. [PMID: 23308270 PMCID: PMC3538684 DOI: 10.1371/journal.pone.0053666] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Accepted: 12/03/2012] [Indexed: 02/01/2023] Open
Abstract
Gfi1b (growth factor independence 1b) is a zinc finger transcription factor essential for development of the erythroid and megakaryocytic lineages. To elucidate the mechanism underlying Gfi1b function, potential downstream transcriptional targets were identified by chromatin immunoprecipitation and expression profiling approaches. The combination of these approaches revealed the oncogene meis1, which encodes a homeobox protein, as a direct and prominent target of Gfi1b. Examination of the meis1 promoter sequence revealed multiple Gfi1/1b consensus binding motifs. Distinct regions of the promoter were occupied by Gfi1b and its cofactors LSD1 and CoREST/Rcor1, in erythroid cells but not in the closely related megakaryocyte lineage. Accordingly, Meis1 was significantly upregulated in LSD1 inhibited erythroid cells, but not in megakaryocytes. This lineage specific upregulation in Meis1 expression was accompanied by a parallel increase in di-methyl histone3 lysine4 levels in the Meis1 promoter in LSD1 inhibited, erythroid cells. Meis1 was also substantially upregulated in gfi1b−/− fetal liver cells along with its transcriptional partners Pbx1 and several Hox messages. Elevated Meis1 message levels persisted in gfi1b mutant fetal liver cells differentiated along the erythroid lineage, relative to wild type. However, cells differentiated along the megakaryocytic lineage, exhibited no difference in Meis1 levels between controls and mutants. Transfection experiments further demonstrated specific repression of meis1 promoter driven reporters by wild type Gfi1b but neither by a SNAG domain mutant nor by a DNA binding deficient one, thus confirming direct functional regulation of this promoter by the Gfi1b transcriptional complex. Overall, our results demonstrate direct yet differential regulation of meis1 transcription by Gfi1b in distinct hematopoietic lineages thus revealing it to be a common, albeit lineage specific, target of both Gfi1b and its paralog Gfi1.
Collapse
Affiliation(s)
- Asif H. Chowdhury
- Department of Biology, The City College of New York and The Graduate Center of The City University of New York, New York, New York, United States of America
| | - Johnny R. Ramroop
- Department of Biology, The City College of New York and The Graduate Center of The City University of New York, New York, New York, United States of America
| | - Ghanshyam Upadhyay
- Department of Biology, The City College of New York and The Graduate Center of The City University of New York, New York, New York, United States of America
| | - Ananya Sengupta
- Department of Biology, The City College of New York and The Graduate Center of The City University of New York, New York, New York, United States of America
| | - Anna Andrzejczyk
- Department of Biology, The City College of New York and The Graduate Center of The City University of New York, New York, New York, United States of America
| | - Shireen Saleque
- Department of Biology, The City College of New York and The Graduate Center of The City University of New York, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
142
|
Polychronidou M, Lohmann I. Cell-type specific cis-regulatory networks: insights from Hox transcription factors. Fly (Austin) 2012; 7:13-7. [PMID: 23221502 DOI: 10.4161/fly.22939] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Hox proteins are a prominent class of transcription factors that specify cell and tissue identities in animal embryos. In sharp contrast to tissue-specifically expressed transcription factors, which coordinate regulatory pathways leading to the differentiation of a selected tissue, Hox proteins are active in many different cell types but are nonetheless able to differentially regulate gene expression in a context-dependent manner. This particular feature makes Hox proteins ideal candidates for elucidating the mechanisms employed by transcription factors to achieve tissue-specific functions in multi-cellular organisms. Here we discuss how the recent genome-wide identification and characterization of Hox cis-regulatory elements has provided insight concerning the molecular mechanisms underlying the high spatiotemporal specificity of Hox proteins. In particular, it was shown that Hox transcriptional outputs depend on the cell-type specific interplay of the different Hox proteins with co-regulatory factors as well as with epigenetic modifiers. Based on these observations it becomes clear that cell-type specific approaches are required for dissecting the tissue-specific Hox regulatory code. Identification and comparative analysis of Hox cis-regulatory elements driving target gene expression in different cell types in combination with analyses on how cofactors, epigenetic modifiers and protein-protein interactions mediate context-dependent Hox function will elucidate the mechanistic basis of tissue-specific gene regulation.
Collapse
Affiliation(s)
- Maria Polychronidou
- Centre for Organismal Studies Heidelberg, University of Heidelberg, Heidelberg, Germany.
| | | |
Collapse
|
143
|
Abstract
The homeobox (HOX) genes are a highly conserved family of homeodomain-containing transcription factors that specify cell identity in early development and, subsequently, in a number of adult processes including hematopoiesis. The dysregulation of HOX genes is associated with a number of malignancies including acute myeloid leukemia (AML) and acute lymphoid leukemia (ALL), where they have been shown to support the immortalization of leukemic cells both as chimeric partners in fusion genes and when overexpressed in their wild-type form. This review covers our current understanding of the role of HOX genes in normal hematopoiesis, AML and ALL, with particular emphasis on the similarities and differences of HOX function in these contexts, their hematopoietic downstream gene targets and implications for therapy.
Collapse
|
144
|
|
145
|
Distinct regulation of c-myb gene expression by HoxA9, Meis1 and Pbx proteins in normal hematopoietic progenitors and transformed myeloid cells. Blood Cancer J 2012; 2:e76. [PMID: 22829978 PMCID: PMC3389162 DOI: 10.1038/bcj.2012.20] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Revised: 05/03/2012] [Accepted: 05/07/2012] [Indexed: 12/11/2022] Open
Abstract
The proto-oncogenic protein c-Myb is an essential regulator of hematopoiesis and is frequently deregulated in hematological diseases such as lymphoma and leukemia. To gain insight into the mechanisms underlying the aberrant expression of c-Myb in myeloid leukemia, we analyzed and compared c-myb gene transcriptional regulation using two cell lines modeling normal hematopoietic progenitor cells (HPCs) and transformed myelomonocytic blasts. We report that the transcription factors HoxA9, Meis1, Pbx1 and Pbx2 bind in vivo to the c-myb locus and maintain its expression through different mechanisms in HPCs and leukemic cells. Our analysis also points to a critical role for Pbx2 in deregulating c-myb expression in murine myeloid cells cotransformed by the cooperative activity of HoxA9 and Meis1. This effect is associated with an intronic positioning of epigenetic marks and RNA polymerase II binding in the orthologous region of a previously described alternative promoter for c-myb. Taken together, our results could provide a first hint to explain the abnormal expression of c-myb in leukemic cells.
Collapse
|
146
|
Akalin A, Garrett-Bakelman FE, Kormaksson M, Busuttil J, Zhang L, Khrebtukova I, Milne TA, Huang Y, Biswas D, Hess JL, Allis CD, Roeder RG, Valk PJM, Löwenberg B, Delwel R, Fernandez HF, Paietta E, Tallman MS, Schroth GP, Mason CE, Melnick A, Figueroa ME. Base-pair resolution DNA methylation sequencing reveals profoundly divergent epigenetic landscapes in acute myeloid leukemia. PLoS Genet 2012; 8:e1002781. [PMID: 22737091 PMCID: PMC3380828 DOI: 10.1371/journal.pgen.1002781] [Citation(s) in RCA: 237] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2011] [Accepted: 05/04/2012] [Indexed: 11/18/2022] Open
Abstract
We have developed an enhanced form of reduced representation bisulfite sequencing with extended genomic coverage, which resulted in greater capture of DNA methylation information of regions lying outside of traditional CpG islands. Applying this method to primary human bone marrow specimens from patients with Acute Myelogeneous Leukemia (AML), we demonstrated that genetically distinct AML subtypes display diametrically opposed DNA methylation patterns. As compared to normal controls, we observed widespread hypermethylation in IDH mutant AMLs, preferentially targeting promoter regions and CpG islands neighboring the transcription start sites of genes. In contrast, AMLs harboring translocations affecting the MLL gene displayed extensive loss of methylation of an almost mutually exclusive set of CpGs, which instead affected introns and distal intergenic CpG islands and shores. When analyzed in conjunction with gene expression profiles, it became apparent that these specific patterns of DNA methylation result in differing roles in gene expression regulation. However, despite this subtype-specific DNA methylation patterning, a much smaller set of CpG sites are consistently affected in both AML subtypes. Most CpG sites in this common core of aberrantly methylated CpGs were hypermethylated in both AML subtypes. Therefore, aberrant DNA methylation patterns in AML do not occur in a stereotypical manner but rather are highly specific and associated with specific driving genetic lesions. Acute myeloid leukemias (AML) are a group of malignancies that originate in the bone marrow. While many different genetic lesions have been linked to the different forms of this disease, it is also clear that these genetic lesions are not always sufficient to cause AML. DNA methylation plays a role in gene expression regulation, and abnormal distribution of DNA methylation has been observed in many cancers, including AML. Here we demonstrate that changes in DNA methylation in AML are not uniform across all AML subtypes, but rather they display unique patterns, which are closely linked to the underlying genetic lesions of each of the different forms of AML. Furthermore, these unique patterns of DNA methylation have different impacts on gene expression regulation in each AML subtype.
Collapse
Affiliation(s)
- Altuna Akalin
- Department of Physiology and Biophysics and the HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medical College, New York, New York, United States of America
| | - Francine E. Garrett-Bakelman
- Department of Medicine, Division of Hematology/Oncology, Weill Cornell Medical College, New York, New York, United States of America
| | - Matthias Kormaksson
- Department of Public Health, Weill Cornell Medical College, New York, New York, United States of America
| | - Jennifer Busuttil
- Department of Medicine, Division of Hematology/Oncology, Weill Cornell Medical College, New York, New York, United States of America
| | - Lu Zhang
- Illumina, Hayward, California, United States of America
| | | | - Thomas A. Milne
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Yongsheng Huang
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Debabrata Biswas
- Laboratory of Chromatin Biology, The Rockefeller University, New York, New York, United States of America
| | - Jay L. Hess
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - C. David Allis
- Laboratory of Chromatin Biology, The Rockefeller University, New York, New York, United States of America
| | - Robert G. Roeder
- Laboratory of Molecular Biology and Biochemistry, The Rockefeller University, New York, New York, United States of America
| | - Peter J. M. Valk
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Bob Löwenberg
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Ruud Delwel
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Hugo F. Fernandez
- Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
| | - Elisabeth Paietta
- Cancer Center, Montefiore Medical Center–North Division, Bronx, New York, United States of America
| | - Martin S. Tallman
- Leukemia Service, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | | | - Christopher E. Mason
- Department of Physiology and Biophysics and the HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medical College, New York, New York, United States of America
- * E-mail: (CEM); (AM); (MEF)
| | - Ari Melnick
- Department of Medicine, Division of Hematology/Oncology, Weill Cornell Medical College, New York, New York, United States of America
- Department of Pharmacology, Weill Cornell Medical College, New York, New York, United States of America
- * E-mail: (CEM); (AM); (MEF)
| | - Maria E. Figueroa
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, United States of America
- * E-mail: (CEM); (AM); (MEF)
| |
Collapse
|
147
|
Breitinger C, Maethner E, Garcia-Cuellar MP, Schambony A, Fischer KD, Schilling K, Slany RK. HOX genes regulate Rac1 activity in hematopoietic cells through control of Vav2 expression. Leukemia 2012; 27:236-8. [PMID: 22713647 DOI: 10.1038/leu.2012.166] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
148
|
He M, Chen P, Arnovitz S, Li Y, Huang H, Neilly MB, Wei M, Rowley JD, Chen J, Li Z. Two isoforms of HOXA9 function differently but work synergistically in human MLL-rearranged leukemia. Blood Cells Mol Dis 2012; 49:102-6. [PMID: 22633751 DOI: 10.1016/j.bcmd.2012.05.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Accepted: 05/01/2012] [Indexed: 02/07/2023]
Abstract
HOXA9 plays a critical role in both normal hematopoiesis and leukemogenesis, particularly in the development and maintenance of mixed lineage leukemia (MLL)-rearranged leukemia. Through reverse transcription-polymerase chain reaction (RT-PCR) analysis of HOXA9 transcripts in human leukemia and normal bone marrow samples, we identified a truncated isoform of HOXA9, namely HOXA9T, and found that both HOXA9T and canonical HOXA9 were highly expressed in leukemia cell lines bearing MLL rearrangements, relative to human normal bone marrow cells or other subtypes of leukemia cells. A frameshift in HOXA9T in exon I causes a premature stop codon upstream of the PBX-binding domain and the homeodomain, which leads to the generation of a non-homeodomain-containing protein. Unlike the canonical HOXA9, HOXA9T alone cannot transform normal bone marrow progenitor cells. Moreover, HOXA9T cannot cooperate with MEIS1 to transform cells, despite the presence of a MEIS1-binding domain. Remarkably, although the truncated isoforms of many proteins function as dominant-negative competitors or inhibitors of their full-length counterparts, this is not the case for HOXA9T; instead, HOXA9T synergized with HOXA9 in transforming mouse normal bone marrow progenitor cells through promoting self-renewal and proliferation of the cells. Collectively, our data indicate that both truncated and full-length forms of HOXA9 are highly expressed in human MLL-rearranged leukemia, and the truncated isoform of HOXA9 might also play an oncogenic role by cooperating with canonical HOXA9 in cell transformation and leukemogenesis.
Collapse
Affiliation(s)
- Miao He
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|